Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
PLoS One ; 18(2): e0276578, 2023.
Article in English | MEDLINE | ID: covidwho-2227485

ABSTRACT

Effective measures are needed to prevent the spread and infectivity of SARS-CoV-2 that causes COVID-19. Chemical inactivation may help to prevent the spread and transmission of this and other viruses. Hence, we tested the SARS-CoV-2 antiviral activity of acetic acid, the main component of vinegar, in vitro. Inactivation and binding assays suggest that acetic acid is virucidal. We found that 6% acetic acid, a concentration typically found in white distilled vinegar, effectively inactivated SARS-CoV-2 after 15-min incubation with a complete loss of replication of competent virus as measured by TCID50. Transmission electron microscopy further demonstrated that 6% acetic acid disrupts SARS-CoV-2 virion structure. In addition, 6% acetic acid significantly inhibits and disrupts the binding of SARS-CoV-2 spike protein binding to ACE2, the primary SARS-CoV-2 cell receptor, after contact with spike protein for 5, 10, 30 and 60 minutes incubation. Taken together, our findings demonstrate that acetic acid possesses inactivating activity against SARS-CoV-2 and may represent a safe alternative to commonly used chemical disinfectants to effectively control the spread of SARS-CoV-2.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/physiology , Acetic Acid/pharmacology , Angiotensin-Converting Enzyme 2/chemistry , Spike Glycoprotein, Coronavirus/chemistry
2.
Viruses ; 15(1)2022 Dec 30.
Article in English | MEDLINE | ID: covidwho-2227484

ABSTRACT

Increasing evidence suggests that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection impacts neurological function both acutely and chronically, even in the absence of pronounced respiratory distress. Developing clinically relevant laboratory mouse models of the neuropathogenesis of SARS-CoV-2 infection is an important step toward elucidating the underlying mechanisms of SARS-CoV-2-induced neurological dysfunction. Although various transgenic models and viral delivery methods have been used to study the infection potential of SARS-CoV-2 in mice, the use of commonly available laboratory mice would facilitate the study of SARS-CoV-2 neuropathology. Herein we show neuroinflammatory profiles of immunologically intact mice, C57BL/6J and BALB/c, as well as immunodeficient (Rag2-/-) mice, to a mouse-adapted strain of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2 (MA10)). Our findings indicate that brain IL-6 levels are significantly higher in BALB/c male mice infected with SARS-CoV-2 MA10. Additionally, blood-brain barrier integrity, as measured by the vascular tight junction protein claudin-5, was reduced by SARS-CoV-2 MA10 infection in all three strains. Brain glial fibrillary acidic protein (GFAP) mRNA was also elevated in male C57BL/6J infected mice compared with the mock group. Lastly, immune-vascular effects of SARS-CoV-2 (MA10), as measured by H&E scores, demonstrate an increase in perivascular lymphocyte cuffing (PLC) at 30 days post-infection among infected female BALB/c mice with a significant increase in PLC over time only in SARS-CoV-2 MA10) infected mice. Our study is the first to demonstrate that SARS-CoV-2 (MA10) infection induces neuroinflammation in laboratory mice and could be used as a novel model to study SARS-CoV-2-mediated cerebrovascular pathology.


Subject(s)
COVID-19 , SARS-CoV-2 , Mice , Male , Female , Animals , COVID-19/pathology , Lung , Neuroinflammatory Diseases , Mice, Inbred C57BL , Disease Models, Animal , Mice, Transgenic
3.
Alzheimer's & dementia : the journal of the Alzheimer's Association ; 18(Suppl 4), 2022.
Article in English | EuropePMC | ID: covidwho-2218666

ABSTRACT

Background COVID‐19 survivors develop neuro‐psychological symptoms as post‐COVID 19 syndromes, although the pathophysiology remains elusive. SARS‐CoV‐2 MA10 (MA10) is a model of mouse‐adapted SARS‐CoV‐2 that recapitulates acute respiratory distress syndrome observed in humans. However, the consequence of MA10 infection on brain pathology is not yet evaluated. The study aimed to evaluate the long‐term consequence of MA10 infection on blood‐brain barrier (BBB) integrity and neuroinflammation Method 10‐week and 12‐months old female BALB/c/cAnNHsd mice were infected intranasally with 104 PFU and 103 PFU SARS‐CoV‐2 MA 10 respectively. The brain was isolated 60‐ and 120‐days post‐infection (60 and 120 dpi) and subjected to histological analysis and mRNA expression by qPCR array. Result Immunohistochemical analysis showed that MA 10 infection caused increased GFAP immunoreactivity in young mice. Histopathological analysis showed that MA10 infection induced significant neuropathology. Ongoing quantification on microglial activation and BBB integrity elucidated a possible correlation between infection and neuropathology. Conclusion Our data shows the first time that MA10 infection induces neuropathological outcomes several weeks after infection. These observations can help to develop novel therapeutic strategies to ameliorate neuroinflammation and restore brain function

4.
Clin Infect Dis ; 75(1): e1195-e1201, 2022 Aug 24.
Article in English | MEDLINE | ID: covidwho-2017769

ABSTRACT

The relationship between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) dose, infection, and coronavirus disease 2019 (COVID-19) outcomes remains poorly understood. This review summarizes the existing literature regarding this issue, identifies gaps in current knowledge, and suggests opportunities for future research. In humans, host characteristics, including age, sex, comorbidities, smoking, and pregnancy, are associated with severe COVID-19. Similarly, in animals, host factors are strong determinants of disease severity, although most animal infection models manifest clinically with mild to moderate respiratory disease. The influence of variants of concern as it relates to infectious dose, consequence of overall pathogenicity, and disease outcome in dose-response remains unknown. Epidemiologic data suggest a dose-response relationship for infection contrasting with limited and inconsistent surrogate-based evidence between dose and disease severity. Recommendations include the design of future infection studies in animal models to investigate inoculating dose on outcomes and the use of better proxies for dose in human epidemiology studies.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Comorbidity , Female , Humans , Pregnancy
6.
Frontiers in cellular and infection microbiology ; 12, 2022.
Article in English | EuropePMC | ID: covidwho-1863930
7.
Nat Commun ; 13(1): 1745, 2022 04 01.
Article in English | MEDLINE | ID: covidwho-1773978

ABSTRACT

Neurological manifestations are a significant complication of coronavirus disease (COVID-19), but underlying mechanisms aren't well understood. The development of animal models that recapitulate the neuropathological findings of autopsied brain tissue from patients who died from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are critical for elucidating the neuropathogenesis of infection and disease. Here, we show neuroinflammation, microhemorrhages, brain hypoxia, and neuropathology that is consistent with hypoxic-ischemic injury in SARS-CoV-2 infected non-human primates (NHPs), including evidence of neuron degeneration and apoptosis. Importantly, this is seen among infected animals that do not develop severe respiratory disease, which may provide insight into neurological symptoms associated with "long COVID". Sparse virus is detected in brain endothelial cells but does not associate with the severity of central nervous system (CNS) injury. We anticipate our findings will advance our current understanding of the neuropathogenesis of SARS-CoV-2 infection and demonstrate SARS-CoV-2 infected NHPs are a highly relevant animal model for investigating COVID-19 neuropathogenesis among human subjects.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Brain , Endothelial Cells , Humans , Primates
8.
Life Sci ; 284: 119881, 2021 Nov 01.
Article in English | MEDLINE | ID: covidwho-1347741

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an infectious disease that has spread worldwide. Current treatments are limited in both availability and efficacy, such that improving our understanding of the factors that facilitate infection is urgently needed to more effectively treat infected individuals and to curb the pandemic. We and others have previously demonstrated the significance of interactions between the SARS-CoV-2 spike protein, integrin α5ß1, and human ACE2 to facilitate viral entry into host cells in vitro. We previously found that inhibition of integrin α5ß1 by the clinically validated small peptide ATN-161 inhibits these spike protein interactions and cell infection in vitro. In continuation with our previous findings, here we have further evaluated the therapeutic potential of ATN-161 on SARS-CoV-2 infection in k18-hACE2 transgenic (SARS-CoV-2 susceptible) mice in vivo. We discovered that treatment with single or repeated intravenous doses of ATN-161 (1 mg/kg) within 48 h after intranasal inoculation with SARS-CoV-2 lead to a reduction of lung viral load, viral immunofluorescence, and improved lung histology in a majority of mice 72 h post-infection. Furthermore, ATN-161 reduced SARS-CoV-2-induced increased expression of lung integrin α5 and αv (an α5-related integrin that has also been implicated in SARS-CoV-2 interactions) as well as the C-X-C motif chemokine ligand 10 (Cxcl10), further supporting the potential involvement of these integrins, and the anti-inflammatory potential of ATN-161, respectively, in SARS-CoV-2 infection. To the best of our knowledge, this is the first study demonstrating the potential therapeutic efficacy of targeting integrin α5ß1 in SARS-CoV-2 infection in vivo and supports the development of ATN-161 as a novel SARS-CoV-2 therapy.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19 Drug Treatment , COVID-19/prevention & control , Oligopeptides/therapeutic use , SARS-CoV-2/physiology , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , COVID-19/virology , Genome, Viral , Humans , Integrins/metabolism , Liver/enzymology , Liver/pathology , Lung/pathology , Lung/virology , Male , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/pharmacology , SARS-CoV-2/genetics , Staining and Labeling , Viral Load/genetics
9.
Cytokine Growth Factor Rev ; 58: 1-15, 2021 04.
Article in English | MEDLINE | ID: covidwho-1101169

ABSTRACT

SARS-CoV-2 is a novel coronavirus that severely affects the respiratory system, is the cause of the COVID-19 pandemic, and is projected to result in the deaths of 2 million people worldwide. Recent reports suggest that SARS-CoV-2 also affects the central nervous system along with other organs. COVID-19-associated complications are observed in older people with underlying neurological conditions like stroke, Alzheimer's disease, and Parkinson's disease. Hence, we discuss SARS-CoV-2 viral replication and its inflammation-mediated infection. This review also focuses on COVID-19 associated neurological complications in individuals with those complications as well as other groups of people. Finally, we also briefly discuss the current therapies available to treat patients, as well as ongoing available treatments and vaccines for effective cures with a special focus on the therapeutic potential of a small 5 amino acid peptide (PHSCN), ATN-161, that inhibits SARS-CoV-2 spike protein binding to both integrin α5ß1 and α5ß1/hACE2.


Subject(s)
COVID-19/complications , Nervous System Diseases/virology , Neurogenic Inflammation/virology , SARS-CoV-2/pathogenicity , Age Factors , Aged , Aged, 80 and over , COVID-19/epidemiology , Humans , Nervous System Diseases/epidemiology , Neurogenic Inflammation/complications , Neuroimmunomodulation/physiology , Pandemics
10.
JACC Basic Transl Sci ; 5(11): 1124-1126, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-893977
11.
JACC Basic Transl Sci ; 6(1): 1-8, 2021 Jan.
Article in English | MEDLINE | ID: covidwho-866798

ABSTRACT

Many efforts to design and screen therapeutics for the current severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic have focused on inhibiting viral host cell entry by disrupting angiotensin-converting enzyme-2 (ACE2) binding with the SARS-CoV-2 spike protein. This work focuses on the potential to inhibit SARS-CoV-2 entry through a hypothesized α5ß1 integrin-based mechanism and indicates that inhibiting the spike protein interaction with α5ß1 integrin (+/- ACE2) and the interaction between α5ß1 integrin and ACE2 using a novel molecule (ATN-161) represents a promising approach to treat coronavirus disease-19.

SELECTION OF CITATIONS
SEARCH DETAIL